+ All Categories
Home > Documents > Ge Seminar

Ge Seminar

Date post: 05-Apr-2018
Category:
Upload: prasad-phapale
View: 215 times
Download: 0 times
Share this document with a friend

of 107

Transcript
  • 7/31/2019 Ge Seminar

    1/107

    PurifyingChallenging ProteinsPrinciples and Methods

    GE Healthcare

  • 7/31/2019 Ge Seminar

    2/107

  • 7/31/2019 Ge Seminar

    3/107

    Purifying

    Challenging ProteinsPrinciples and Methods

  • 7/31/2019 Ge Seminar

    4/107

    2 Handbook 28-9095-31 AA

    Content

    Introduction...............................................................................................................................................................5

    Challenging proteins ........................................................................................................................................................5

    Outline ....................................................................................................................................................................................5

    Common acronyms and abbreviations ..................................................................................................................6

    Symbols .................................................................................................................................................................................7

    Chapter 1Membrane proteins ..........................................................................................................................................................9

    Introduction..........................................................................................................................................................................9

    Classification of membrane proteins ....................................................................................................................10

    Purification of integral membrane proteins for structural and functional studies .........................11

    Starting material .............................................................................................................................................................12

    Small-scale expression screening of histidine-tagged membrane proteins

    from E. coli lysates .........................................................................................................................................................15

    Cell harvest ......................................................................................................................................................................17

    Cell disruption and membrane preparation ......................................................................................................18

    Solubilization ....................................................................................................................................................................20

    Purification ........................................................................................................................................................................29

    Purification of non-tagged membrane proteins .............................................................................................38

    Purity and homogeneity check ...............................................................................................................................40

    Conditioning ......................................................................................................................................................................43

    Proteomic analysis of membrane proteins ........................................................................................................45

    References .........................................................................................................................................................................49

    Chapter 2Multiprotein complexes ...............................................................................................................................................51

    Introduction.......................................................................................................................................................................51

    Pull-down assays ...........................................................................................................................................................53

    Isolation of native complexes ...................................................................................................................................61

    Isolation of recombinant protein complexes ....................................................................................................63

    References .........................................................................................................................................................................67

    Chapter 3Inclusion bodies ..............................................................................................................................................................69

    Optimizing for soluble expression ..........................................................................................................................69

    Strategies for handling inclusion bodies .............................................................................................................70Isolation of inclusion bodies ......................................................................................................................................71

    Solubilization ....................................................................................................................................................................72

  • 7/31/2019 Ge Seminar

    5/107

    Handbook 28-9095-31 AA 3

    Refolding ...........................................................................................................................................................................72

    Refolding using gel filtration .....................................................................................................................................74

    Analysis of refolding .....................................................................................................................................................82

    References .........................................................................................................................................................................82

    Appendix 1Principles and standard conditions for different purification techniques ...........................................83

    Affinity chromatography (AC) ...................................................................................................................................83

    Ion exchange chromatography (IEX) ....................................................................................................................84

    Hydrophobic interaction chromatography (HIC) ............................................................................................86

    Gel filtration (GF) chromatography ........................................................................................................................87

    Reversed phase chromatography (RPC) .............................................................................................................88

    Appendix 2Manual and automated purification .....................................................................................................................89

    Tagged recombinant proteins for simple purification ..................................................................................89

    Manual purification techniques ...............................................................................................................................89

    Automated purification using KTAdesign chromatography systems ................................................90

    Appendix 3Column packing and preparation .........................................................................................................................93

    Column selection ............................................................................................................................................................95

    Appendix 4Conversion data: proteins, column pressures ..........................................................................................................96

    Column pressures ..........................................................................................................................................................96

    Appendix 5...................................................................................................................................................................97Converting from linear flow (cm/h) to volumetric flow rates (ml/min) and vice versa .....................97

    Appendix 6Amino acids table ...........................................................................................................................................................98

    Related literature.............................................................................................................................................. 100Ordering information .................................................................................................................................... 101

  • 7/31/2019 Ge Seminar

    6/107

    Handbook 28-9095-31 AA 5

    IntroductionThis handbook is intended for students and experienced researchers with an interest in the

    isolation of integral membrane proteins, multiprotein complexes, or in refolding proteins from

    inclusion bodies. The aim is to present tools, strategies, and solutions available to meet the

    purification challenges associated with these three classes of proteins.

    For a background on techniques for protein purification in general and handling recombinant

    proteins, we recommend the Recombinant Protein Purification Handbook and other

    handbooks in this series (see Related literature on page 100).

    Challenging proteinsOur knowledge and understanding of the structural and functional biology of soluble proteins

    has increased dramatically over the last decade. Much of the technology for the production,

    purification, and analysis of soluble proteins is now at a stage where generic purification

    protocols allow relatively high success rates.

    The situation is different for the areas that this handbook covers; integral membrane proteins,multiprotein complexes, and inclusion bodies. The need to handle and study these more

    difficult groups of proteins is clear, given that:

    about 30% of a typical cells proteins are membrane proteins, and more than 50% of the

    current drugs on the market exert their actions via membrane proteins

    while carrying out their enzymatic, structural, transporting, or regulatory functions,

    proteins most often interact with each other, forming multiprotein complexes

    a large proportion of normally soluble proteins that are overexpressed in E. coli end up as

    incorrectly folded and insoluble protein in inclusion bodies

    OutlineAfter a general introduction to each area, high-level consensus workflows are presented to

    summarize current best practices in each area. Rather than providing a number of detailed

    protocols that have been optimized for individual proteins, this handbook provides general

    advice or generic protocols in a step-by-step format. The generic protocols are intended

    as starting points for development of separation protocols. Details will typically have to be

    changed to tailor the protocols for individual proteins. Furthermore, the required variations

    to the generic protocols cannot be predicted and unless appropriate changes are made, the

    protocols will only work poorly, if at allthis is one of the major challenges for the researcher

    involved with these groups of proteins. To address this issue, the generic protocols are

    presented with critical parameters identified, together with ranges of values to test for the

    parameters. The handbook also provides guidance, hints, and tips when using protocols other

    than those described here.

  • 7/31/2019 Ge Seminar

    7/107

    6 Handbook 28-9095-31 AA

    Common acronyms and abbreviations

    LTAB lauryl trimethylammoniumbromide

    MBP maltose binding protein

    MPa megaPascalM

    rrelative molecular weight

    MS mass spectrometryN/m column efficiency expressed

    as theoretical plates per

    meterPBS phosphate buffered salinepI isoelectric point, the pH

    at which a protein has zeronet surface charge

    PMSF phenylmethylsulfonyl fluoridepsi pounds per square inchPVDF polyvinylidene fluoride

    r recombinant, as in rGSTRNase ribonucleaseRPC reversed phase

    chromatographySDS sodium dodecyl sulfateSDS-PAGE sodium dodecyl sulfate

    polyacrylamide gelelectrophoresis

    TAP Tandem affinity purification

    TCEP Tris (2-carboxyethyl)phosphine hydrochloride

    TEV Tobacco etch virusu units (e.g., of an enzyme)Y2H Yeast-two-hybrid

    A280

    absorbance at specifiedwavelength (in thisexample, 280 nanometers)

    AC affinity chromatographyBCA bicinchoninic acid

    CBP calmodulin binding peptideCDNB 1-chloro-2-4-dinitrobenzeneC. elegans Caenorhabditis elegans

    CF chromatofocusingCHAPS 3-[(3-cholamidopropyl)

    dimethylammonio]-

    1-propanesulfonateCMC critical micellar concentration

    CV column volumeDAB 3,3-diaminobenzidineDDM dodecyl maltoside

    DNase deoxyribonucleaseDS desalting (sometimes referred

    to as buffer exchange)

    DTT dithiothreitolE. coli Escherichia coliELISA enzyme-linked

    immunosorbent assayFF Fast Flow

    FW formula weight

    GF gel filtration (sometimesreferred to as SEC: size

    exclusion chromatography)GFP green fluorescent proteinGPCR G-protein coupled receptor

    GSH reduced glutathioneGSSG oxidized glutathione

    GST glutathione-S-transferaseGua-HCl guanidine-HClHIC hydrophobic interaction

    chromatographyHMW high molecular weightHP High Performance

    HRP horseradish peroxidaseIEX ion exchange

    chromatography (also seen asIEC in the literature)

    IMAC immobilized metal ion affinity

    chromatographyIPTG isopropyl -D-thiogalactoside

    LDAO lauryldimethylamine oxideLMW low molecular weight

  • 7/31/2019 Ge Seminar

    8/107

    Handbook 28-9095-31 AA 7

    Symbols

    This symbol indicates general advice to improve procedures or recommend action

    under specific situations.

    This symbol denotes mandatory advice and gives a warning when special care should

    be taken.

    This symbol highlights troubleshooting advice to help analyze and resolve difficulties.

    Yellow highlights indicate chemicals, buffers, and equipment

    Blue highlights indicate an experimental protocol

  • 7/31/2019 Ge Seminar

    9/107

    Handbook 28-9095-31 AA 9

    Chapter 1Membrane proteins

    IntroductionMembrane proteins play key roles in fundamental biological processes, such as transport ofmolecules, signaling, energy utilization, and maintenance of cell and tissue structures. About

    30% of the genes determined by genome sequencing encode membrane proteins, and these

    proteins comprise more than 50% of the current drug targets. Despite their importance, our

    knowledge of the structure and function of membrane proteins at the molecular level lags far

    behind that for soluble proteins. For instance, at the time of the publication of this handbook,

    membrane proteins only represent around 1% of the 3-D atomic resolution structures that

    have been deposited in the Protein Data Bank (http://www.pdb.org/).

    Integral membrane proteins exist in a lipid environment of biological membranes

    (biomembrane), but the available techniques for purifying, handling, and analyzing them

    were optimized for water-soluble proteins in an aqueous environment. To be able to handle

    and study membrane proteins they must be dispersed in an aqueous solution. This is usually

    accomplished by adding a detergent that solubilizes the biomembrane and forms a soluble

    complex with the lipids and membrane proteins (Fig 1.1). Solubilization is a harsh treatment

    that has to be carefully optimized to avoid protein loss and inactivation. Protein denaturation

    and/or aggregation are frequently encountered. Solubilization is one of the most critical

    aspects in handling membrane proteins.

    Other difficulties contribute to our lack of detailed structural and functional understanding of

    membrane proteins. These include:

    Low abundance. The quantity of membrane proteins is often very low in their naturalsetting. This makes their natural source impractical as a starting material for their

    preparation.

    Difficult production. Heterologous overexpression often results in low expression levels

    and inactive protein due to insufficient membrane insertion and folding or lack of post-

    translational modifications. Over-expression of membrane proteins can be toxic to the cell.

    Fig 1.1. Schematic drawing of detergent solubilization of membrane proteins. Membrane proteins are transferred fromthe natural lipid bilayer (blue and yellow) to complexes with detergent (green) and, in some cases, lipids. A lipid-detergentmicelle, a detergent micelle, and free detergent are also shown.

  • 7/31/2019 Ge Seminar

    10/107

    10 Handbook 28-9095-31 AA

    Membrane protein expression, purification, and analysis present considerable challenges.

    Nevertheless, a substantial number of membrane proteins, especially from bacterial origin,

    have been over-produced, isolated, and characterized in molecular detail. Also, several

    studies aiming at mapping the membrane proteome in different organisms have been

    published. Due to great efforts in a number of membrane protein labs, generic protocols for

    membrane protein work have begun to emerge. These protocols are extremely useful as a

    starting point in the lab. In the main part of this chapter, such protocols are provided togetherwith optimization advice and references for further reading.

    Classification of membrane proteinsMembrane proteins are classified as peripheral or integral. Peripheral membrane proteins are

    loosely associated with the membrane and are usually water soluble after being released

    from the membrane. Peripheral membrane proteins generally present limited methodological

    challenges when compared with integral membrane proteins. Throughout this handbook, the

    term membrane protein refers to integral membrane protein unless otherwise indicated.

    Integral membrane proteins are insoluble in water. They have one or more transmembrane

    segments comprising polypeptide stretches that span the membrane. The transmembranemoiety can be constituted of a single or a bundle of-helices or of-barrel-like structures

    composed of multiple polypeptide stretches. These proteins are called -helical membrane

    proteins (Fig 1.2, left) and -barrel membrane proteins (Fig 1.2, right), respectively. The

    -barrel membrane proteins are predominant in the outer membrane of Gram-negative

    bacteria and mitochondria. Some proteins display both structures.

    Fig 1.2. Three dimensional structure representations of an -helical membrane protein (left;Anabaena sensoryrhodopsin; PDB ID: 1XIO; [1]) and a -barrel membrane protein (right; ferric hydroxamate uptake receptor (fhua) fromE. coli; PDB ID: 1FCP; [2]). The structures are oriented such that the externally exposed area of each protein is at thetop. The yellow lines show the approximate locations of the lipid bilayer membrane. The yellow, horizontal lines are for

    illustration purposes only and are not based on crystallographic data. Structures from The Protein Data Bank(http://www.pdb.org/).

  • 7/31/2019 Ge Seminar

    11/107

    Handbook 28-9095-31 AA 11

    Purification of integral membrane proteins for structural andfunctional studiesThe high level workflow for the production and purification of integral membrane proteins

    for structural and functional studies is shown in Figure 1.3. Each of the different stages in the

    workflow is described in detail below, with relevant protocols. The protocols are intended as

    starting points in the lab. Hints, tips, useful variations, and troubleshooting advice are also

    given. The focus is on protocols for bacterial membrane production and purification sincethis is most common. Protocols for eukaryotic membrane proteins are less well developed.

    However, much of the general advice is also valid for work with eukaryotic membrane

    proteins.

    Fig 1.3. Workflow overview for membrane protein isolation and purification for structural and functional studies.

    Natural source Cloning and expression

    Cell harvest

    Expression screening

    Cell disruption andmembrane prep

    Solubilization

    Purification

    Purity andhomogeneity check

    Conditioning

    Structural and/orfunctional studies

    Detergent screening

  • 7/31/2019 Ge Seminar

    12/107

    12 Handbook 28-9095-31 AA

    Starting materialMembrane proteins from natural sourcesThe natural source of a membrane protein can be considered as a starting material for

    purification. The only three-dimensional structure in molecular detail that has been reported

    to date for a eukaryotic G-protein coupled receptor (GPCR), bovine rhodopsin, was obtained

    with protein purified from bovine retina, where the protein is highly abundant (8). In many

    cases, however, low abundance of the target protein precludes the use of the natural source

    as starting material.

    Examples of purifications from natural sources are presented later in this chapter.

    CloningVectors used for the expression of soluble proteins are also commonly used for the

    production of membrane proteins. It is useful to design a number (10 to 50) of different

    constructs, including different homologues, to increase the chance that a particular

    membrane protein can be produced in an active form.

    In addition to the general considerations for choosing a vector (see Recombinant ProteinPurification Handbook, see Related literature, page 100), a number of other aspects relate

    more specifically to choosing a vector for expressing membrane proteins.

    Affinity tagging greatly facilitates expression screening based on chromatographic

    enrichment, as well as optimization and use of protocols for purification of membrane

    proteins. Polyhistidine tags are commonly used for membrane proteins, but the GST-

    tag and others have also been used successfully. The insertion of a protease cleavage

    site between the affinity tag and the target protein enables removal of the tag before

    further analyses.

    While a hexahistidine tag (His6) is the standard option for water-soluble proteins,longer histidine tags (with 8 or 10 histidine residues) are often used for membrane

    proteins to increase the binding strength and thus improve yields in IMAC purification.

    Drawbacks with longer (> 6 histidine residues) histidine tags are that expression

    levels have been reported to be decreased in some cases and that a higher imidazole

    concentration is required for elution.

    Tags should generally be placed on the C-terminal end of the protein to reduce risk of

    affecting the membrane insertion process based on the N-terminal signal peptide.

    Fusion of the target membrane protein to a fluorescent protein tag such as GFP

    in combination with a histidine tag allows direct and convenient visualization ofthe target during expression, solubilization, and purification and can speed up the

    optimization of these processes (6).

  • 7/31/2019 Ge Seminar

    13/107

    Handbook 28-9095-31 AA 13

    Expression and screeningTo correctly decide which conditions and constructs will be best suited for producing the

    protein for the intended study, an efficient screening protocol is essential. Because of the

    relatively low concentrations of overexpressed membrane proteins, it is useful to apply

    affinity tags combined with separation methods that allow enrichment of the target protein.

    Overexpression is a major bottleneck in the overall workflow for membrane protein

    production. Several host systems are available and the final choice will depend both on

    protein-specific requirements (e.g., for post-translational modifications) and practical aspects

    (e.g., available equipment in the lab and expertise). It is often useful to try different hosts or

    host strains in parallel for a particular target protein to increase the likelihood of success. In

    addition, homologous membrane proteins from several sources can be cloned in parallel to

    be able to select those that express well.

    E. coli strain BL21 (DE3) is the most commonly used host for overexpression of membrane

    proteins, in combination with a pET vector. High expression levels for functional membrane

    proteins are usually more than an order of magnitude lower than for overexpression of

    water-soluble proteins in E. coli. One inherent issue is that membrane proteins need to beinserted into membranes, and the availability of membrane structures in most cells is limited.

    The issue with limited membrane availability can be addressed by using a host with large

    amounts of internal membranes (e.g., Rhodobacter spp.; (3)). Another way of avoiding the

    limitations set by available membranes is to produce the membrane protein as inclusion

    bodies. This is usually not desired but may allow preparation of active protein through

    solubilization of the inclusion bodies using denaturants followed by refolding. Successful

    refolding of-barrel membrane proteins from inclusion bodies has been achieved (4) but

    refolding of-helical membrane proteins is an even greater challenge. For a separate

    discussion on inclusion bodies, see chapter 3.

    A modest growth and expression rate is beneficial to avoid the formation of inclusion

    bodies when using E. coli as a host. This can be achieved by the use of a weak

    promoter, a low concentration of inducer and/or lowering the growth temperature

    after induction.

    An overview of different expression systems for membrane proteins is provided in Table 1.1.

    For a review on important considerations for membrane protein expression, see reference 7.

  • 7/31/2019 Ge Seminar

    14/107

    14 Handbook 28-9095-31 AA

    Table 1.1. Overexpression systems used for prokaryotic and eukaryotic membrane protein production

    Expression system Advantages Disadvantages

    E. coli The most widely usedoverexpression system for(prokaryotic) membrane protein

    production.

    Often not suitable for overexpression ofeukaryotic membrane proteins

    No glycosylation and limited post-translational modifications

    Yeast Can perform some post-translational modifications

    Several different yeast systemshave been used for membraneprotein production (5)

    Does not produce high cell densities(S. cerevisiae)

    Hyperglycosylation can occur(S. cerevisiae)

    Different lipids (compared with mammaliancells)

    Insect cells Less complex growth

    conditions compared withmammalian cells

    Relatively high expressionlevels

    Glycosylation

    More costly and complex than E. coli or

    yeast; different lipids (compared withmammalian cells)

    Mammalian cells CHO, BHK and other cell linesare often used for functionalstudies of receptors

    Authentic (mammalian) proteinis produced

    High cost and complex work

    Rhodobacterspp. High expression levels throughcoordinated synthesis offoreign membrane proteinswith synthesis of new internalmembranes (3)

    Different lipids (compared with mammaliancells)

    Cell free Allows expression of toxicproteins and proteins that areeasily degraded in vivo

    Allows incorporation of labeledand non-natural amino acids.

    High cost

    Membrane protein insertion in membraneor detergent micelle has not been fullydeveloped

    Disposable 96-well filter plates, from GE Healthcare, prepacked with affinity purification

    media for histidine- or GST-tagged proteins can be used for reproducible, high-throughput

    screening of protein expression. Typical applications include expression screening of different

    constructs, screening for suitable detergents and solubility of proteins, and optimization of

    the conditions for small-scale parallel purification. Plates are available prepacked with Ni

    SepharoseTM High Performance or Ni Sepharose 6 Fast Flow for working with histidine-tagged

    proteins (His MultiTrapTM HP or His MultiTrap FF, respectively); and Glutathione Sepharose 4Fast Flow or Glutathione Sepharose 4B for working with GST-tagged proteins (GST MultiTrap

    FF or GST MultiTrap 4B, respectively).

  • 7/31/2019 Ge Seminar

    15/107

    Handbook 28-9095-31 AA 15

    Small-scale expression screening of histidine-tagged membraneproteins from E. coli lysates

    Cell lysis and solubilization

    Buffer preparation

    Lysis buffer: 20 mM sodium phosphate, 100 mM NaCl, 2 mM MgCl2, 20 mM imidazole, 0.5 mM Tris(2-carboxyethyl) phosphine hydrochloride (TCEP), 5 U/ml benzonase,1 mg/ml lysozyme, EDTA-free protease inhibitor cocktail, (concentration accordingto manufacturers recommendation),1-2% of a selection of detergents, pH 7.4

    Procedure

    1. Harvest cells from the culture by centrifugation at 7000 to 8000 g for 10 min or at 1000 to1500 g for 30 min at 4C.

    2. Discard the supernatant. Place the bacterial pellet on ice.

    3. Suspend the bacterial pellet by adding 5 to 10 ml of lysis buffer for each gram of wet cells.To prevent the binding of host cell proteins with exposed histidines, it is essential to includeimidazole at a low concentration in the sample and binding buffer.

    4. Leave for 2 h with mild agitation at room temperature or 4C, depending on the sensitivity ofthe target protein.

    5. Measure and adjust pH if needed.

    Expression screening procedure

    Materials

    His MultiTrap HP or His MultiTrap FF

    Centrifuge that handles 96-well plates

    Buffer preparation

    Binding buffer: 20 mM sodium phosphate, 500 mM NaCl, 20 to 40 mM imidazole, 0.5 mM TCEP,1 to 2% detergent, pH 7.4. (The optimal imidazole concentration is protein

    dependent; 20 to 40 mM is suitable for many proteins.)

    Wash buffer: 20 mM sodium phosphate, 500 mM NaCl, 20 to 40 mM imidazole, 0.5 mM TCEP,0.03% dodecyl maltoside (DDM), 1 to 2% detergent, pH 7.4,

    Elution buffer: 20 mM sodium phosphate, 500 mM NaCl, 500 mM imidazole, 0.5 mM TCEP, 0.03%DDM, 1 to 2% detergent, pH 7.4

    To increase the purity, use as high a concentration of imidazole as possible in the

    sample and binding buffers without losing binding capacity.

  • 7/31/2019 Ge Seminar

    16/107

    16 Handbook 28-9095-31 AA

    Preparing the filter plate

    1. Peel off the bottom seal from the 96-well filter plate. Be sure to hold the filter plate over a sinkto accommodate any leakage of storage solution when removing the bottom seal.

    2. Hold the filter plate upside down and gently shake it to dislodge any medium adhering to thetop seal. Return the filter plate to an upright position.

    3. Place the filter plate against the bench surface and peel off the top seal.

    4. Position the filter plate on top of a collection plate.

    Note: Remember to change or empty the collection plate as necessary during the followingsteps.

    5. Centrifuge the filter plate for 2 min at 200 g to remove the ethanol storage solution from themedium.

    6. Add 500 l of deionized water to each well. Centrifuge the plate for 2 min at 200 g.

    7. Add 500 l of binding buffer to each well to equilibrate the medium. Centrifuge for 2 min at200 g. Repeat once. The filter plate is now ready for use.

    Blank run: Reducing agents may be used in sample and buffers. If this is the case,replace step 7 with the following steps:

    7. Add 500 l of elution buffer/well. No reducing agent should be used in the elutionbuffer during this blank run. Centrifuge the plate for 2 min at 200 g

    8. Add 500 l of binding buffer including reducing agent to each well to equilibratethe medium. Centrifuge for 2 min at 200 g. Repeat once. The filter plate is now

    ready for use with reducing agent. Do not store His MultiTrap plates with buffers

    containing reducing agents.

    Centrifugation procedureCheck that all wells are drained after centrifugation. If not, then increase the

    centrifugation force slightly.

    Do not apply a force of more than 700 g during centrifugation.

    1. Apply 100 l of lysate to each well of the filter plate and incubate for 3 min.

    Note: If the yield of protein is very low, increase the incubation time and/or gently agitate thefilter plate to mix. The lysate volume can also be increased, and several aliquots of lysate canbe added successively to each well.

    2. Centrifuge the plate at 100 g for 4 min or until all the wells are empty. Discard theflowthrough.

    3. Add 50 l of binding buffer per well. Centrifuge at 200 g for 2 min.

    4. Add 200 l of wash buffer per well. Centrifuge at 200 g for 2 min. Repeat twice.

    5. Add 50 l of elution buffer per well and mix for 1 min.

    Note: The volume of elution buffer can be varied (50 to 600 l per well), depending on theconcentration of target protein required.

    6. Change the collection plate and centrifuge at 200 g for 2 min to collect the eluted protein.Repeat twice or until all the target protein has been eluted (A

    280should be < 0.1, indicating that

    all protein has been eluted).

    Note: If necessary, change the collection plate between each elution to prevent unnecessarydilution of the target protein.

  • 7/31/2019 Ge Seminar

    17/107

    Handbook 28-9095-31 AA 17

    The following detergents have been used with this protocol: 1% FOS-Choline 12,

    1% undecyl maltoside, 1% dodecyl maltoside, 1% Cymal-5, 1% Cymal-6, 2% octyl

    glucoside, 1% Triton X-100, 1% lauryl dimethylamine oxide (LDAO).

    To optimize the protocol, vary the concentration of imidazole in the sample and in the

    binding and wash buffers. A common variation range with His MultiTrap plates is 20 to

    40 mM imidazole. If binding of the target protein is too low with these concentrations,

    try 5 to 20 mM imidazole. In general, too low of an imidazole concentration in the

    binding and wash buffers can cause adsorption of unwanted host proteins (and hence

    a lower purity). Too high of an imidazole concentration can lead to a reduced yield of

    the target protein.

    Analysis

    Samples can by analyzed by SDS-PAGE with Coomassie Blue staining (see Purity and

    homogeneity check on page 40), or by dot-blot analysis on nitrocellulose membrane.

    Histidine-tagged proteins can be detected using Anti-His Antibody.

    Cell harvestMethods for cell harvest are host dependent and the same protocols are used for the

    recovery of membrane proteins as for intracellular water-soluble proteins. Cell harvest of

    suspension cultures is done by low speed centrifugation.

    Cell harvest ofE. coli cultures

    Buffer preparation

    PBS: 10 mM phosphate, 2.7 mM KCl, 137 mM NaCl, pH 7.4

    Centrifugation procedure

    1. Centrifuge at 6000 to 9000 g at 4C for 15 min to collect the cells. Discard the supernatant.Resuspend the cells in 500 ml ice-cold PBS.

    2. Centrifuge at 6000 to 9000 g at 4C for 15 min. Discard the supernatant. Resuspend the cellpellet in 10 ml ice-cold PBS, or another volume as required.

    3. The resuspended cell pellet can be stored at 80C.

  • 7/31/2019 Ge Seminar

    18/107

    18 Handbook 28-9095-31 AA

    Cell disruption and membrane preparationMethods for cell disruption are host dependent and essentially the same protocols are used

    for recovery of membrane proteins as for water-soluble proteins. Cell disruption yields a

    suspension of membrane fragments/vesicles that contains the membrane proteins. The

    suspension also contains soluble proteins, remaining intact cells, various cell debris, and

    other material as contaminants. These contaminants may need to be removed, depending on

    the purification procedure used. Differential centrifugation is the standard approach for theisolation of membrane fragments/vesicles after cell disruption.

    The pellet from cell harvest is resuspended in a suitable buffer for cell disruption (e.g., PBS).

    DNase is added to reduce viscosity. It is useful to add a protease inhibitor cocktail to reduce

    possible protein degradation. A selection of commonly used techniques for cell disruption are

    summarized in Table 1.2.

    Table 1.2. Overview of techniques for cell disruption to yield a suspension of membrane vesicles

    Technique Principle Advantages (+) / Disadvanatges (-)

    Liquid shear pressure(e.g., French press)

    Rapid pressure drop by transferringthe sample from a chamber at highpressure through an orifice into achamber at low pressure

    + Fast and efficient, also for largevolumes

    - Causes heating of the sample (cooling isrequired)

    Ultrasonication Cells disrupted by high frequencysound

    + Simple- Causes heating of the sample, which

    can be difficult to control by cooling- Proteins may be destroyed by shearing- Noisy

    - Not for large volumes

    Glass bead milling Agitation of the cells with fine glassbeads

    + Useful for cells that are more difficult todisrupt (e.g., yeast)

    - Somewhat slow and noisy

    Osmotic shock Change from high to low osmoticmedium

    + Simple, inexpensive- Only useful for disruption of cells with

    less robust walls (e.g., animal cells)

    Repeated freezing andthawing

    Cells disrupted by repeatedformation of ice crystals; usuallycombined with enzymatic lysis

    + Simple, inexpensive+ Yields large membrane fragments- Slow

    - May damage sensitive proteins anddissociate membrane protein complexes

    - Low yield

    Enzymatic lysis Often used in combination withother techniques, e.g., freeze-thawing or osmotic shock;lysozyme is commonly used tobreak cell walls of bacteria

    + Gentle+ Yields large membrane fragments- Slow- Low yield

  • 7/31/2019 Ge Seminar

    19/107

    Handbook 28-9095-31 AA 19

    Cell disruption from frozen E. coli cell paste with overexpressed membraneprotein

    Solutions

    PBS: 10 mM phosphate, 2.7 mM KCl, 137 mM NaCl, pH 7.4

    MgCl2: 1 M

    Pefabloc: 100 mM

    DNase: 20 mg/ml

    Lysozyme: 10 mg/ml

    Cell disruption

    1. For each gram of cell paste combine, from the above solutions, 5 ml PBS, 5 l MgCl2, 50 l

    Pefabloc, 5 l DNase, and 80 l lysozyme. Mix until the suspension is homogenous.

    2. Sonicate on ice. Use the manufacturers recommended settings for amplitude and time for the

    probe being used (e.g. 5 min of accumulated time; 9 s on, 5 s off).

    3. Continue immediately with the membrane preparation.

    This procedure was modified from reference 6.

    Membrane preparation should be performed immediately after cell disruption.

    Membrane preparation from E. coli.

    All steps are carried out at 4C or on ice.

    1. Centrifuge at 24 000 g for 12 min. Collect the supernatant.

    2. Centrifuge the supernatant at 150 000 g for 45 min. Remove the supernatant and resuspendthe pellet in 10 ml PBS. The pellet contains the membrane fraction.

    3. Centrifuge the resuspended pellet at 150 000 g for 45 min. Remove the supernatant andresuspend the pellet in 5 ml PBS.

    4. Determine protein concentration using standard methods for soluble proteins such as theBiuret method or the bicinchoninic acid (BCA) method.

    5. For storage, rapidly freeze the membrane suspension dropwise using liquid nitrogen and storeat 80C.

    This procedure was taken from reference 6.

    Different cell disruption protocols may give rise to different size fragments; the

    centrifugation speed needs to be optimized accordingly.

    Water crystals formed upon slow freezing may harm membrane proteins. Fast

    freezing by submersion of the membrane suspension in liquid nitrogen forms

    amorphous ice structures thus reducing the negative effects of freezing (some

    researchers avoid freezing completely and always perform membrane protein

    preparation from cell to pure protein as fast as possible, without interruptions; see

    next point).

  • 7/31/2019 Ge Seminar

    20/107

    20 Handbook 28-9095-31 AA

    For unstable membrane proteins, it may be beneficial to proceed directly with

    purification after the preparation of membranes, and thus avoid freezing and storing

    the membranes.

    For small scale (0.5 to 50 ml) membrane preparations from E. coli, a procedure with

    lysozyme treatment followed by water lysis and centrifugation is often efficient .

    To facilitate protein purification, it can be useful to separate the inner and outermembranes from E. coli membrane preparations. This can be particularly helpful for

    large (1 to >10 l) preparations. The inner membrane can be selectively solubilized

    with 2% N-lauroylsarcosine (9). The outer membranes can then be recovered in

    the pellet after a 1 h centrifugation. An alternative is to separate the inner and

    outer membranes by a long (~10 h) sucrose gradient centrifugation, following cell

    disruption.

    It is sometimes possible to omit the fairly lengthy and cumbersome membrane preparation

    step. The alternative is to first disrupt the cells and then directly solubilize membrane proteins

    by the addition of detergent to the cell lysate, with no prior isolation of membranes. The

    resulting solubilisate can be used for chromatography directly. By using chromatographycolumns that accept direct loading of unclarified homogenized cell lysate and detergent-

    treated unclarified lysate (e.g., HisTrapTM FF crude columns), histidine-tagged membrane

    proteins can be purified directly from the cell lysate (see Purification of histidine-tagged

    membrane protein directly from crude, solubilized E. coli lysate, page 33).

    SolubilizationThis is one of the most critical stages during the preparation of membrane proteins. During

    the solubilization stage, membrane proteins are extracted from their natural environment,

    the lipid membrane, to an aqueous environment by the use of detergents. Detergents act by

    disintegrating the lipid bilayer while incorporating lipids and proteins in detergent micelles.

    The hydrophobic surface areas of the membrane proteins and the lipid tails are buried in

    the hydrophobic interior of the detergent micellar structures, while hydrophilic parts of the

    proteins are in contact with the aqueous environment (Fig 1.1). An efficient solubilization

    dissociates most lipid-protein and protein-protein interactions, thereby allowing the

    separation of proteins.

    The target protein can be purified in the presence of detergent by applying essentially any

    of the existing protein purification techniques available for soluble proteins. A successful

    solubilization protocol extracts the membrane protein at a high yield and results in stable

    protein-detergent complexes (or protein-lipid-detergent complexes) where the protein retainsits active conformation.

    Some membrane proteins require the interaction with native lipids from the lipid bilayer or

    added exogenous lipids to remain in their active conformation. In such cases, it is essential

    that the solubilization protocol enables the formation of a stable protein-lipid-detergent

    complex and that it does not remove the required native lipid(s) associated with the target

    protein. Harsh solubilization and purification procedures may lead to the removal of such

    essential lipids, and hence inactivation of the protein.

  • 7/31/2019 Ge Seminar

    21/107

    Handbook 28-9095-31 AA 21

    Detergents and CMCDetergents are amphipathic substances with a polar (hydrophilic) head group and a nonpolar,

    (hydrophobic) tail. The polar part can be nonionic, anionic, cationic, or zwitterionic and

    detergents are often classified accordingly (i.e., a detergent with an anionic polar part is

    referred to as an anionic detergent). Advantages and disadvantages of the different classes of

    detergents are listed in Table 1.3.

    Table 1.3. Advantages and disadvantages with the different classes of detergents

    Detergent class Advantages Disadvantages

    Nonionic(e.g., dodecyl maltoside)

    Generally mild and non-denaturingWidely used

    May give low solubilization yields

    Ionic (anionic or cationic)(e.g., SDS, LTAB)

    Can be extremely efficient insolubilization

    Often denaturingInterfere with ion exchangeseparations

    Zwitterionic(e.g., FOS-Choline 12)

    Often used in membrane proteincrystallization.Combines the advantages of ionic andnon-ionic detergents

    More denaturing than nonionicdetergents

    Above a certain concentration in an aqueous environment, detergent molecules associate

    to form multimolecular complexes, micelles, with hydrophobic interiors and hydrophilic

    surfaces. This concentration is referred to as the critical micellar concentration (CMC). The

    CMC is different for different detergents and it also varies with pH, temperature and ionic

    strength.

    In general, all buffers and solutions used for membrane protein preparations (for

    solubilization, purification, storage, etc.) should have a detergent concentration above

    the CMC.

    Several detergents that are recommended for solubilization of membrane proteins are listed

    in Table 1.4. Chemical structures of a few example detergents from the different classes are

    shown in Figure 1.4.

  • 7/31/2019 Ge Seminar

    22/107

    22 Handbook 28-9095-31 AA

    Table 1.4. Some recommended detergents for solubilization of membrane proteins

    Detergent Class1 FWCMC2(mM)

    Brij 35 N 1200 0.07

    C12E8 N 539 0.11

    CHAPS (3[(3-Cholamidopropyl) dimethylammonio]propanesulfonic acid)

    Z 615 8

    Cymal 7 (Cyclohexyl-n-heptyl--D-maltoside)

    N 522 0.19

    Decyl maltoside N 483 1.8

    Digitonin N 1229 < 0.5

    DDM (dodecyl maltoside) N 511 0.17

    FOS-Choline 12 Z 352 0.12Hecameg (6-O-(N-Heptylcarbamoyl)methyl--D-gluco pyranoside)

    N 335 19.5

    LDAO (lauryldimethylamine oxide) Z 229 1

    Nonidet P40 N 615 0.25

    Nonyl glucoside N 306 6.5

    Octyl glucoside N 292 18

    Tween 20 N 1228 0.06

    Triton X-100 N 647 0.23

    1 N = nonionic; Z = zwitterionic2 At 20C to 25C and ~50 mM Na+

  • 7/31/2019 Ge Seminar

    23/107

    Handbook 28-9095-31 AA 23

    Fig 1.4. Chemical structures of selected detergents from the different classes.

    Detergent screeningSeveral different detergents and conditions should be screened to establish the best

    conditions for each membrane protein. Solubilization yield is monitored by assaying for the

    target membrane protein in the solubilized fraction. In the protocol below, non-solubilized

    material is removed by ultracentrifugation.

    It is also possible to apply non-clarified solubilisate directly to specially designed

    chromatography columns or multiplates that can handle cellular debris (see Expression

    screening of histidine-tagged membrane proteins from E. coli lysates). Filtration can also be

    used for clarification.

    Protein detection may be performed by Western blot after SDS-PAGE. For low-expressed

    proteins, a chromatographic enrichment step may be required before protein detection is

    possible (see Conditioning on page 43). For highly expressed proteins, solubilization yield can

    be estimated by SDS-PAGE with Coomassie Blue staining.

    O O-

    Na+

    O

    O

    S

    HO

    HO

    HO HO

    O

    O

    N+

    O-

    Ionic: Sodium dodecyl sulfate

    Non-ionic: Octyl glucoside

    Zwitterionic: LDAO (Lauryl dimethylamine oxide)

  • 7/31/2019 Ge Seminar

    24/107

    24 Handbook 28-9095-31 AA

    In addition to determination of yield and homogeneity, it is often necessary to monitor

    protein activity, which is the best indication of intact protein structure (and function). For

    some membrane proteins, functional assays can be applied to the detergent-solubilized

    protein. In other cases, reinsertion of the membrane protein into an artificial lipid bilayer

    (membrane protein reconstitution) is necessary to perform a functional assay.

    General detergent screening procedureBuffer preparation

    PBS: 10 mM phosphate, 2.7 mM KCl, 137 mM NaCl, pH 7.4

    Tris buffer: 20 mM at neutral pH

    Table 1.5. Additives to PBS or Tris buffer for detergent screening

    Membrane proteinconcentration(mg/ml)

    Detergentconcentration (%) NaCl (mM) Temp(C) Time

    Start condition 5 1(always above CMC)

    100 4 2 h

    Variation range 1 to 10 0.4 to 2(always above CMC)

    100 to 500 4 to 37 5 min 5 h

    For a list of recommended detergents, see Table 1.4.

    Solubilization

    1. Starting with the membrane preparation of known protein concentration, combine materialsas shown in Table 1.5 to give a total volume of 1 ml. Incubate with gentle mixing at the desiredtemperature for the time indicated.

    2. Centrifuge at 100 000 g at 4C for 45 min.

    3. Assay the supernatant for target protein. Generic assays that can be used are SDS-PAGE (seePurity and homogeneity check on page 40), rapid affinity purification followed by gel filtration(see Purity and homogeneity check on page 41) and affinity tag specific assays (e.g., a dot blotusing tag-specific antibodies). Also, assays for membrane protein activity should be considered.

    The expression screening protocol on page 15, Small-scale expression screening

    of histidine-tagged membrane proteins from E. coli lysates can be adapted to a

    combined solubilization screening and binding screening protocol.

    The results from a preliminary solubilization screening of EM29, an Mr29 000 histidine-tagged

    membrane protein from E. coli, is shown in Fig 1.5. A strong band at the expected position for

    the target protein indicates high expression and/or efficient solubilization. FC12, Triton X-100

    and LDAO gave weaker bands than the other tested detergents. It was thus concluded that

    these detergents were less suitable for solubilization of this protein. It should be stated alsothat high or reasonable yields must be combined with preservation of activity and stability

    in detergent solution after solubilizationwhich is the next screening analysis that should be

    considered.

  • 7/31/2019 Ge Seminar

    25/107

    Handbook 28-9095-31 AA 25

    Fig 1.5. Analysis by SDS-PAGE and Coomassie Blue staining of a combined expression screening and preliminarysolubilization screening of EM29. Eight different detergents were tested and elution was done in two steps (left and

    right half of the gel, respectively). FC12 = FOS-Choline 12; UDM = undecyl maltoside; DDM = dodceyl maltoside; OG =octyl glucoside; TX-100 = Triton X-100; LDAO = lauryl dimethylamine oxide. Data kindly provided by Dr. Said Eshaghi,Karolinska Insitute, Stockholm, Sweden.

    Specific lipids may be associated with the protein in the native membrane, and their

    presence can be essential for protein activity. Retaining activity requires that these

    lipids are still present after solubilization and purification. Harsh solubilization and

    purification conditions can lead to removal of such lipids, resulting in inactivation of

    the membrane protein.

    DDM is often a good detergent to try in initial solubilization tests.CHAPS and digitonin have been reported to work particularly well for solubilization of

    membrane proteins from Pichia pastoris.

    FC12 DDM Cymal6 TX-100 FC12 DDM Cymal6 TX-100

    UDM Cymal5 OG LDAO UDM Cymal5 OG LDAO

    Mr 103

    97

    66

    45

    30

    20.1

    14.4

    EM29

  • 7/31/2019 Ge Seminar

    26/107

    26 Handbook 28-9095-31 AA

    Solubilization screening for GST-tagged membrane proteins produced in E. coliThe method described below (Fig 1.6) provides a simple and rapid method to select for

    optimal solubilization conditions to obtain the highest yield of GST-tagged membrane protein.

    This screening procedure is based on the affinity of GST for Glutathione Sepharose 4B media.

    The detergents selected for the screening must not affect the GST-binding activity

    Fig 1.6. Detergent screening assay for a GST-tagged membrane protein.

    Materials

    GST Detection Module

    GST SpinTrapTM Purification Module

    GST MultiTrap 4B

    Binding buffer: 10 mM phosphate, 2.7 mM KCl, 137 mM NaCl, pH 7.4 (PBS)Elution buffer: Binding buffer supplemented with 0.2% (w/v) detergent and 10 mM reduced

    glutathione

    1

    Determine the detergenteffect on the enzymaticactivity of purified GSTwith the CDNB assay

    Solubilize membranes indifferent detergents andconcentrations that donot affect the activity ofthe GST tag

    Purify fusion protein using GSTSpinTrap Purification Module

    Analyze yield by SDS-PAGE andactivity of membrane protein

    (if assay is available)

    96-well microplate

    2

    3

  • 7/31/2019 Ge Seminar

    27/107

    Handbook 28-9095-31 AA 27

    Solubilization screening

    If the enzymatic activity of GST (as an indicator of GST binding activity) in the

    presence of the selected detergents is unknown, evaluate GST activity in the

    detergents using the CDNB assay provided in the GST Detection Module. Compare

    GST activity in the presence and absence of each detergent. GST is known to be fully

    active in DDM, CHAPS, octyl glucoside, Tween 20, Triton X-100, Brij35 and NP 40 at

    detergent concentrations between 0.3 to 10 times CMC for each detergent .

    1. Disrupt cells by lysozyme treatment combined with freeze-thawing and isolate membranes bycentrifugation at ~50,000 g for 30 min at 4C (see Membrane preparation from E. coli on page19 for additional information). Higher speed and longer centrifugation times (e.g., 100 000 gfor 60 min) may be required if more harsh cell disruption methods are used.

    2. Solubilize the membrane pellet (1:10 w/v) in 5% (w/v) detergent solutions for 2 h on ice withmild agitation (see Detergent screening on page 23 for additional information).

    3. Clarify by centrifugation at ~50 000 g for 30 min at 4C (or at 18 000g for 60 min at 4C).

    4. Decant 500 l of the supernatant and purify with GST SpinTrap Purification Module or GSTMultiTrap 4Baccording to the supplied instructions. Elute with 500 l elution buffer. Assay forGST activity with GST Detection Module.

    5. Analyze by SDS-PAGE (see Purity and homogeneity check on page 40).

    This procedure was taken from reference 10.

    Optimization of solubilization conditionsAfter establishing the initial conditions for solubilization, the conditions can be optimized by

    further screening with one or a limited number of detergents. Useful screening parameters to

    investigate are: ratio of protein concentration to detergent concentration. Protein concentrations in the

    range of 1 to 10 mg/ml are typical

    solubilization time and mixing conditions (e.g., mild agitation, end-over-end rotation, or

    vigorous stirring)

    pH

    ionic strength

    Size homogeneity can be used as an indicator of stability (and therefore optimum

    solubilization conditions) because membrane proteins often oligomerize or aggregaterapidly when destabilized. Size homogeneity can be rapidly evaluated using SuperdexTM

    200 5/150 GL (see Size homogeneity characterization on page 40). Figure 1.7 demonstrates

    how Superdex 200 5/150 GL (column volume 3 ml) can be used to screen for homogeneity

    under various pH and salt conditions.

  • 7/31/2019 Ge Seminar

    28/107

    28 Handbook 28-9095-31 AA

    Fig 1.7. Screening of pH and ion strength conditions for optimal homogeneity and stability of a detergent-proteincomplex. Rapid gel filtration with Superdex 200 5/150 GL showed a symmetrical peak when the separation wasperformed at pH 5.2 in 0.1 M NaCl (A), indicating a homogenous protein under these conditions. At somewhat highersalt concentration (D) a small peak appeared close to the void volume, indicating that oligomerization or aggregationappeared to a limited extent . At both pH 7.5 and pH 9.5 significant peaks were obtained close to the void volume,indicating severe oligomerization or aggregation. The complete screening procedure was achieved in only a few hours,including the time for column equilibration. Sample consumption was 6 10 l for the complete screen. Data kindly

    provided by Dr. Said Eshagi, Karolinska Institute, Stockholm, Sweden.

    Mixtures of detergents could also be considered.

    Additives such as lipids and small amphiphiles (e.g., 1,2,3-heptanetriol) or known

    ligands for the target protein may be useful to test. Small amphiphiles may affect

    crystallization behavior by changing the size of detergent micelles. Ligands are

    believed to reduce the dynamics of the protein structure and thus stabilize the protein

    in detergent solution.

    Solubilization for purificationAfter optimization, the solubilization protocol is performed at a scale that is appropriate for

    the amount of membrane protein that needs to be obtained.

    It is recommended to proceed with purification immediately after solubilization, to

    minimize the loss of membrane protein activity due to aggregation, loss of structure,

    or proteolytic degradation.

    A detergent that works well for the solubilization of a particular membrane protein

    may be less suited for other operations with the same protein (e.g., crystallization).

    Procedures for exchanging detergents are described in Conditioning (see page 43).

    For reviews on the use of detergents in membrane protein purification, see references 11 and

    12.

    0.0

    5.0

    10.0

    15.0

    20.0

    mAU

    0.0 0.5 1.0 1.5 2.0 2.5 3.0 ml

    : .

    -2.0

    0.0

    2.0

    4.0

    6.0

    8.0

    10.0

    12.0

    mAU

    0.0 0.5 1.0 1.5 2.0 2.5 3.0 ml

    :

    : .

    0.0

    5.0

    10.0

    15.0

    mAU

    0.0 0.5 1.0 1.5 2.0 2.5 3.0 ml

    : .: .

    1.29

    1.76

    0.0

    5.0

    10.0

    15.0

    20.0

    25.0

    30.0

    35.0

    40.0mAU

    0.0 0.5 1.0 1.5 2.0 2.5 3.0 ml

    :

    : .

    0.0

    2.0

    4.0

    6.0

    8.0

    10.0

    12.0

    14.0

    16.0

    mAU

    0.0 0.5 1.0 1.5 2.0 2.5 3.0 ml

    : .

    : .

    1.29

    1.78

    0.0

    2.0

    4.0

    6.0

    8.0

    10.0

    12.0

    14.0mAU

    0.0 0.5 1.0 1.5 2.0 2.5 3.0 ml

    .

    1.3

    : .

    : .

    A B C

    D E F

    0.1 M

    0.3 M

    pH 5.2 pH 7.5 pH 9.5

  • 7/31/2019 Ge Seminar

    29/107

    Handbook 28-9095-31 AA 29

    Solubilization with organic solventsSmall and stable membrane proteins are sometimes possible to extract from the lipid

    bilayer in their active form by the use of organic solvents. For instance, a 110 amino acid

    membrane protein was extracted from E. coli with a chloroform:methanol mixture, and initial

    purification of the extract was done in this environment using an organic solvent resistant

    chromatography medium and column (Fig 1.8; 13). For this membrane protein, a 1:1 ratio of

    chloroform:methanol gave the best separation.

    Fig 1.8. Gel filtration separation of solvent-extracted membrane proteins. Solid line = organic extract ofE. colimembranes in which EmrE (a multidrug resistance transporter protein) was overexpressed; broken line = organicextract of blank E. coli membranes. Two ratios of chloroform (C) and methanol (M) were used. The asterisk marks EmrEcontaining peaks. From reference 13. Used with permission. Copyright 2002 Elsevier Science (USA) Publication.

    PurificationMembrane proteins are usually purified as protein-lipid-detergent complexes. The solubility

    of the complexes in an aqueous environment allow the application of essentially the same

    separation techniques as used for water-soluble proteins. The main difference is that the

    purification of membrane proteins is carried out with detergent present in all solutions. This

    is necessary because protein-detergent complexes are dynamic and would immediatelylose detergent molecules in the absence of free detergent. Detergent concentrations

    should be above the CMC but can be kept about 10 times lower than what was used during

    solubilization (typically in the 0.1% range).

    0

    50

    100

    150

    200

    250

    A280

    (mAU)

    5.0 10.0 15.0 20.0 25.0 30.0 35.0 40.0 ml

    3:1 :M

    1:1 C:M

    Column: Sephadex LH-20 (40 ml bed volume) in 10 mm diameter column.Sample: Organic extract ofE. coli membranes in which EmrE (a multidrug

    resistance transporter protein) was overexpressedEluent: Chloroform (C):methanol (M) 3:1 (upper) and C:M 1:1 (lower).Flow rate: 2 ml/minSystem: KTApurifier

    *

    *

  • 7/31/2019 Ge Seminar

    30/107

    30 Handbook 28-9095-31 AA

    High detergent concentrations can reduce the stability of the protein. However,

    detergent concentrations need to be high during solubilization if the concentration

    of membrane components is high. Once solubilization is completed, detergent

    concentrations can be reduced. Detergents are often expensive, and it is also useful to

    limit consumption for cost reasons.

    Over-purification can lead to the removal of essential lipids from the protein-lipid-

    detergent complex with concomitant loss of protein activity.

    Membrane protein stability can often be improved by having 5% glycerol in all buffers

    throughout the purification.

    Purification of histidine-tagged membrane proteinsHistidine-tagged proteins have affinity for Ni2+ and several other metal ions that can be

    immobilized on chromatographic media using chelating ligands. Consequently, a protein

    containing a histidine tag will be selectively bound to metal-ion-charged media such as Ni

    Sepharose High Performance (HP) and Ni Sepharose 6 Fast Flow (FF) while most other cellular

    proteins will not bind or bind weakly. Elution is achieved by increasing the concentration ofimidazole. This chromatographic technique is often termed immobilized metal ion affinity

    chromatography (IMAC).

    Water-soluble, (histidine)6-tagged proteins are usually straightforward to purify following

    standard protocols. Histidine-tagged membrane proteins are sometimes more problematic,

    and weak binding to IMAC media is often reported. It has been speculated that this is

    due to restricted accessibility of the histidine-tag for the IMAC ligand due to the binding

    of detergent to the protein. To address the issue, longer histidine tags are routinely used

    for overexpression of membrane proteins. Also, the presence of a linker, such as green

    fluorescent protein (GFP), between the histidine tag and the target membrane protein has

    been suggested to improve binding to IMAC media.

    Historically, batch-wise purification has often been employed for the purification of

    histidine-tagged membrane proteins. Batch-wise purification involves mixing the sample

    with the chromatography media in an open vessel for a designated time, often overnight.

    The suspension is then packed into a column for washing and elution of the bound protein.

    Batch-wise purification can sometimes improve yields since adsorption times are longer than

    for column separations. On the other hand, since a batch-wise procedure is longer it also

    leaves the protein more exposed to proteolytic degradation or inactivation, and may thus

    compromise the quality of the purified protein.

    Purification of histidine-tagged membrane proteins can also be performed using column-based methods (see protocols on following pages).

    Depending on the level of purity required for the final application, additional purification

    steps can be performed after IMAC. For this purpose, gel filtration is possibly the most

    common, and has the advantage that optimization is usually not needed. Anion exchange

    chromatography can also be used and is often included between IMAC and gel filtration steps

    (see the section Additional purification steps later in this chapter).

  • 7/31/2019 Ge Seminar

    31/107

    Handbook 28-9095-31 AA 31

    Purification of histidine-tagged membrane protein from the solubilized, isolatedmembrane fraction

    Materials

    Column: HisTrap HP, 1 ml

    Binding buffer: PBS, 20 mM imidazole, 0.1 to 1 % detergent (e.g., DDM), pH 7.4

    Elution buffer: PBS, 0.5 M imidazole, 0.1 to 1% detergent (e.g., DDM), pH 7.4

    Sample preparation

    To 5 ml of solubilized membrane protein, add 0.20 ml of elution buffer to give a final imidazoleconcentration of 20 mM.

    Purification

    This purification procedure should be performed at 4C.

    1. Fill the syringe or pump tubing with distilled water. Remove the stopper and connect thecolumn to the syringe (use the connector supplied), laboratory pump, or chromatographysystem drop to drop to avoid introducing air into the system.

    2. Remove the snap-off end at the column outlet.

    3. Wash out the ethanol with 3 to 5 column volumes (CV) of distilled water.

    4. Equilibrate the column with 10 CV of binding buffer at a flow rate of 1 ml/min*

    5. Apply the sample (using a syringe fitted to the Luer connector or by pumping it onto the

    column) Use a flow rate of 1 ml/min.6. Wash with 10 CV of binding buffer at a flow rate of 1 ml/min.

    7. Elute with a gradient of 0% to 75% elution buffer in 20 CV at a flow rate of 1 ml/min. When asyringe is used, elute stepwise with successively higher concentrations of imidazole.

    8. After elution, wash the column with 5 CV 100% elution buffer followed by 5 CV binding buffer.

    *One ml/min corresponds to approximately 30 drops/min when using a syringe with a HiTrap 1-

    ml column. When using a larger column, a higher flow rate can be used. See column instructions.

    The procedure can be scaled up by connecting two or three columns in series or by

    using HisTrap HP 5 ml columns.

    A relatively low NaCl concentration (e.g., PBS is 150 mM NaCl) is recommended

    because membrane proteins tend to be less soluble at higher ionic strengths.

    Higher concentrations (e.g., 300 to 500 mM NaCl) are often recommended for

    IMAC of water-soluble proteins to reduce ionic interactions of contaminants with

    the chromatographic medium. For some cases, even lower NaCl concentrations

    (e.g., < 150 mM) should be applied for a membrane protein. Alternatively, the NaCl

    concentration can be reduced directly after the IMAC step by desalting the material

    using a HiTrapTM Desalting column.

    It has been reported that by using gradient elution (with increasing concentrationsof imidazole) from an IMAC column, as in the protocol above, protein-lipid-detergent

    complexes that differ only in lipid content can be separated.

  • 7/31/2019 Ge Seminar

    32/107

    32 Handbook 28-9095-31 AA

    Re-application of the flowthrough material, after step 5 above, to allow the sample to

    pass through the IMAC column several times can be useful to maximize yield (14).

    The yield can also be increased by decreasing the flow rate during sample loading.

    For further purification, ion exchange chromatography and/or gel filtration is often

    suitable (see Additional purification steps on page 35).

    Figure 1.9 shows the purification of YedZ-TEV-GFP-(His)8 using the protocol above. YedZ isa transporter membrane protein from E. coli that was overexpressed in E. coli as a fusion

    protein with GFP, a C-terminal (histidine)8

    tag and a tobacco etch virus (TEV) protease

    cleavage sequence.

    Fig 1.9. Two-step purification ofE. coli YedZ-TEV-GFP-(His)8

    from solubilized membranes prepared from E. coli cell culture.Fractions from (A) IMAC were further purified by (B) gel filtration. (C) SDS-PAGE analysis of selected fractions shows thepurity of the target protein. Fraction D from the HisTrap HP column was essentially homogeneous. M = molecular weightmarker. Peak E most likely contained light-scattering detergent-lipid aggregates. Data kindly provided by Dr. David Drew,Center for Biomembrane Research, Stockholm University, Stockholm, Sweden.

    50 100 150 200 250 300 350

    500

    67

    0

    125

    192

    250

    0

    1000

    1500

    2000

    2500

    ml

    mAU(280nm)

    ImidazolemM

    200 40 60 80

    500

    0

    1000

    1500

    2000

    2500

    ml

    mAU(280nm)

    A BA

    B

    C

    D

    E

    F

    IMACColumn: HisTrap HP, 5 mlSample: Solubilized membranes with

    YedZ-TEV-GFP-(His)8 fusion proteinSample load: 2.5 mg fusion protein/ml medium (after solubilization

    at 3 mg/ml)Binding buffer: PBS, 0.1% DDM, pH 7.4Elution buffer: Binding buffer with 500 mM imidazole, pH 7.4Wash: 4% B over 20 CV, 425% over 20 CVElution: 50% BFlow rate: Load 0.3-0.5 ml/min, elution 1 ml/minSystem: KTAprimeTM

    Gel filtrationColumn: Superdex 200 10/300 GLSample: Eluted fractions from HisTrap HPSample load: 0.5 mlBuffer: PBS, 0.1% DDM, pH 7.4Flow rate: 0.4 ml/minSystem: KTAprime

    Totalm

    emb

    ranes

    A B C D E FMr ( 103)

    25015010075

    50

    37

    25

    20

    C

    M

    YedZ-TEV-GFP-(His)8

  • 7/31/2019 Ge Seminar

    33/107

    Handbook 28-9095-31 AA 33

    Purification of histidine-tagged membrane protein directly from crude,solubilized E. coli lysateThis protocol circumvents the need for membrane preparation and centrifugations. It uses

    a chromatography column that was designed for the application of unclarified crude cell

    lysate.

    Material

    Column: HisTrap FF crude, 1 ml

    Binding buffer: PBS, 40 mM imidazole, 0.1% detergent (e.g., DDM), pH 7.4

    Elution buffer: PBS, 1 M imidazole, 0.1% detergent (e.g., DDM), pH 7.4

    Sample preparation

    Suspend E. coli cell paste by addition of 5 ml PBS with 40 mM imidazole for each gram of paste.Perform cell lysis by lysozyme treatment and sonication (see Table 1.2). Add detergent to theunclarified lysate from a concentrated stock (e.g., DDM to a final concentration of 0.8%). Stir on ice

    for 1.5 hours.

    Purification

    1. Fill the pump tubing or syringe with distilled water. Remove the stopper and connect thecolumn to the chromatography system tubing, syringe (use the provided Luer connector), orlaboratory pump drop to drop to avoid introducing air into the system.

    2. Remove the snap-off end at the column outlet.

    3. Wash out the ethanol with 3 to 5 CV of distilled water.

    4. Equilibrate the column with 10 CV of binding buffer at a flow rate of 1 ml/min*

    5. Apply the detergent-treated, unclarified lysate with a pump (0.5 ml/min) or syringe. Loadingvolumes of unclarified lysate are highly dependent on each specific sample.

    *One ml/min corresponds to approximately 30 drops/min when using a syringe with a HiTrap1-ml column. When using a larger column, a higher flow rate can be used. See column

    instructions.

    Continuous, gentle stirring of the sample during sample loading is recommended

    to prevent sedimentation. Sample loading at 4C may increase the viscosity of the

    sample. An adverse effect of increased sample viscosity is that maximum back

    pressure for the column is reached at a lower sample volume loading on the column.

    Large volumes may increase back pressure, making the use of a syringe more

    difficult.

    6. Depending on the sample volume (larger sample volumes require larger wash volumes), washwith 10 to 30 CV of binding buffer at a flow rate of 1 ml/min.

    7. Elute with a gradient of 0% to 12% elution buffer in 10 CV followed by 12% to 100% elutionbuffer in 5 to10 CV, all at a flow rate of 1 ml/min. When a syringe is used, elute stepwise withsuccessively higher concentrations of imidazole.

    8. After elution, wash the column with 5 CV elution buffer followed by 5 CV binding buffer.

    Figure 1.10 shows the purification of YedZ-TEV-GFP-(His)8

    using the protocol above.

  • 7/31/2019 Ge Seminar

    34/107

    34 Handbook 28-9095-31 AA

    For further purification, ion exchange chromatography and gel filtration is often

    suitable (see Additional purification steps on page 35).

    The procedure can be scaled up by connecting two or three columns in series or by

    using HisTrap FF crude 5 ml columns.

    Fig 1.10. Purification of a (histidine)8-tagged membrane protein, YedZ-TEV-GFP-(His)

    8directly from crude, solubilized E.

    coli lysate using HisTrap FF crude, 1 ml. Peak fractions 14 and 15 were analyzed by SDS-PAGE. The gel was scanned todetect the GFP portion of the fusion protein. An arrow indicates the band corresponding to YedZ-TEV-GFP-(His)

    8. In the

    chromatogram, blue =A280

    ; orange = A425

    (to detect YedZ); green = A485

    (to detect GFP); gray = conductivity; red = % elutionbuffer. The overexpression vector was kindly provided by Dr J.-W. deGier, Centre for Biomembrane Research, Stockholm,Sweden.

    800

    200

    400

    600

    mAU

    00

    20

    40

    60

    80

    % Elution buffer

    80.0 85.0 90.0 95.0

    14 15

    100.0 ml

    YedZ-TEV-GFP-(His)8

    1514

    Column: HisTrap FF crude, 1 mlSample: E. coli cell lysate, with overexpressed YedZ-TEV-GFP-(His)8 solubilized

    in 0.8% dodecyl maltoside for 100 min on ice.Sample load: 50 mlBinding buffer: PBS, 40 mM imidazole, 0.1% DDM, pH 7.4Elution buffer: PBS, 1 M imidazole, 0.1% DDM, pH 7.4Flow rate: 1 ml/minGradient: 4% to 12% elution buffer in 10 ml; 12% to 100% elution buffer in 5 mlSystem: KTAexplorer

  • 7/31/2019 Ge Seminar

    35/107

    Handbook 28-9095-31 AA 35

    Additional purification stepsFurther purification of the IMAC-purified, histidine-tagged membrane protein is usually

    necessary for applications that require highly pure homogeneous material (e.g., for structural

    characterizations). One or two additional chromatographic steps are usually sufficient. For

    maximum efficiency, the purification scheme should be designed so that different separation

    principles are utilized in the different steps.

    A highly efficient purification scheme may consist of IMAC (separation according to specific

    affinity) followed by desalting and ion exchange (separation according to charge differences)

    and finally by gel filtration (separation according to size differences). Gel filtration is most

    often used as the last step for the final removal of aggregates and for transfer of the sample

    to a buffer suitable for further functional and structural studies. The separations are carried

    out in the presence of detergent above the CMC (typically about 0.1%). Other conditions

    are the same as for water-soluble proteins. Some suitable columns are listed in Table 1.6.

    KTAxpress chromatography system can be used to automate multistep purifications

    to produce highly pure proteins with minimum hands-on time. KTAdesignTM systems are

    presented in Appendix 2.

    High-resolution anion exchange can be used for both purification and characterization of

    the charge homogeneity of purified membrane proteins. In most cases the ionic strength of

    samples from IMAC must be reduced before application to ion exchange chromatography. Fig

    1.14 shows charge characterization of an IMAC-purified membrane protein using Mono Q.

  • 7/31/2019 Ge Seminar

    36/107

    36 Handbook 28-9095-31 AA

    Table 1.6. Prepacked ion exchange and gel filtration columns for the additional purification of membrane proteins afterinitial IMAC purification

    Technique Column Columnvolume

    (ml)

    Comment

    Anion exchange Mono Q 5/50 GL 1 For the highest resolution and purity in

    anion exchange chromatography at mgscale

    Mini Q 0.24 or 0.8 For the highest resolution and purity inanion exchange chromatography at gscale

    RESOURCE Q 1 or 6 A strong anion exchange medium; for highthroughput and easy scale up

    HiTrap Q HP 1 or 5 A strong anion exchange medium; for fastseparations with high resolution and sharppeaks

    HiLoad 16/10 QSepharose HP 20 Same as previous, but for larger sampleamounts

    HiLoad 26/10 QSepharose HP

    53 Same as previous, but for larger sampleamounts

    HiTrap Q FF 1 or 5 A strong anion exchange medium; for fastseparations with good resolution

    HiPrep 16/10 Q FF 20 Same as the previous, but for largersample amounts

    HiTrap DEAE FF 1 or 5 Weak anion exchange medium; analternative to the strong Q anion

    exchangers

    HiPrep 16/10 DEAE FF 20 Same as the previous, but for largersample amounts

    Cation exchange Mono S 5/50 GL 1 For the highest resolution and purity incation exchange chromatography at mgscale

    Mini S 0.24 or 0.8 For the highest resolution and purity incation exchange chromatography at gscale

    RESOURCE S 1 or 6 A strong cation exchange medium; for

    high throughput and easy scale up

    HiTrap SP HP 1 or 5 A cation exchange medium; for fastseparations with high resolution and sharppeaks

    HiLoad SPSepharose HP

    20 Same as previous, but for larger sampleamounts

    HiLoad SPSepharose HP

    53 Same as previous, but for larger sampleamounts

    HiTrap SP FF 1 or 5 For fast separations with good resolution

    HiPrep 16/10 SP FF 20 Same as the previous, but for largersample amounts

    Table continued next page.

  • 7/31/2019 Ge Seminar

    37/107

    Handbook 28-9095-31 AA 37

    Technique Column Columnvolume

    (ml)

    Comment

    Gel filtration Superdex 200 10/300

    GL

    24 For highest resolution size separations of

    membrane protein-detergent complexesin the molecular weight range 10 000 to600 000* in sample volumes up to 250 l

    HiLoad 16/60Superdex 200 pg

    HiLoad 26/60 Superdex200 pg

    120

    320

    For size separations of proteins in themolecular weight range10 000 to 600 000* in sample volumes upto 5 ml (120 ml column) or 12 ml (320 mlcolumn)

    Superdex 75 10/300 GL 24 For highest resolution size separations of proteins in the molecular weight range3000 -70 000* in sample volumes up to

    250 lHiLoad 16/60 Superdex

    75 pg

    HiLoad 26/60 Superdex75 pg

    120

    320

    For size separations of membrane protein-detergent complexes in the molecularweight range 3000 to 70 000* in samplevolumes up to 5 ml (120 ml column) or 12ml (320 ml column)

    * Determined for soluble, globular proteins.

    The solubility of membrane proteins can be very sensitive to ionic strength. When this

    is the case, fractions eluted in a salt gradient from an ion exchange chromatography

    column should be immediately diluted or run on a PD-10 Desalting, HiTrap Desalting,or a HiPrep 26/10 Desalting column to reduce the salt concentration (e.g., to

    < 50 mM).

    Gel filtration is excellent for the final purification step as it both removes aggregates

    and simultaneously achieves buffer exchange, if required.

    Tag cleavageThe affinity tag can be removed after purification if a protease cleavage site has been

    inserted between the tag and the target protein. If using an KTAxpress system, on-column

    cleavage protocols can be performed automatically. Protease activity can be affected by

    the presence of detergents. Comprehensive data on the activity of different proteases indetergents is largely lacking, but both PreScission Protease and thrombin have been used

    successfully with detergents. It is recommended to check for the extent of cleavage under

    different conditions by SDS-PAGE (see Purity and homogeneity check on page 40).

    PreScission protease exhibits excellent cleavage properties at 4C and is a useful

    alternative for cleavage of sensitive proteins (15).

    TEV protease is fully active in 9 mM decyl maltoside, but is partly inactive in several

    other detergents (16).

    See ordering information on page 103 for proteases available from GE Healthcare.

    Table 1.6. continued

  • 7/31/2019 Ge Seminar

    38/107

    38 Handbook 28-9095-31 AA

    Purification of non-tagged membrane proteinsFor abundant membrane proteins, the natural source, rather than a heterologous host, is

    often the best starting material for the purification. Due to the lack of an affinity tag, and

    hence a highly specific initial purification step, a combination of several classical protein

    purification techniques usually has to be employed to obtain sufficient purity. The columns

    listed in Table 1.6 are also commonly used in purification schemes for non-tagged membrane

    proteins, in combination with columns for other chromatography techniques. For anoverview, see the Protein Purification Handbook in this series.

    Ion exchange chromatography is often used as the main purification step for the purification

    of non-tagged membrane proteins. Figure 1.11 shows the anion exchange purification step

    for a decyl maltoside solubilized glucose transporter membrane protein. The purity after this

    step was above 90%.

    Fig 1.11. Purification of the glucose transporter from human erythrocyte membranes (Glut1) by anion exchangechromatography. Erythrocyte membranes were solubilized with decyl maltoside. Glut1 eluted as a sharp peak at100 mM NaCl with a purity greater than 90%. From reference 17. Used with permission. Copyright 2001 ElsevierPublication.

    Avoid using anionic detergents with anion exchange columns, and cationic detergents

    with cation exchange columns.

    As indicated earlier, gel filtration is the ideal final purification step for membrane proteins. It

    is excellent for polishing as it removes aggregates and other impurities that are of a differentsize than the target protein, while simultaneously performing buffer exchange.

    0.0

    1.0

    0.0

    1.0

    0 5 10 15

    Concentration

    NaCl(M)

    Glut1

    Elution volume (ml)

    A280 nm

    Column: HiTrap Q HP, 1 mlSample: Solubilized human erythrocyte membranes

    Start buffer: 10 mM BisTris, 0.5 mM EDTA, 0.2% DM, pH 6.0Gradient: 0 to 500 mM NaCl in 15 CVFlow rate: Load: 0.5 ml/min; elution: 0.25 ml/min

  • 7/31/2019 Ge Seminar

    39/107

    Handbook 28-9095-31 AA 39

    Figure 1.12 shows the use of repeated gel filtration for final purification of the bovine creatine

    transporter from HEK293 cells (18). Membranes were solubilized with decyl maltoside and

    initial purification was performed on a lectin column (wheat germ agglutinin immobilized on

    Sepharose). Initial gel filtration on a Superdex 200 column gave a broad, nonsymmetrical

    peak (Fig 1.12, upper). Additional gel filtration of a narrow fraction on the same column gave a

    symmetrical peak, indicating homogeneity. Purity was confirmed by SDS-PAGE.

    Fig 1.12.Purification of the bovine creatine transporter from HEK293 cells. Membranes were solubilized with decylmaltoside and initial purification was done on immobilized wheat germ agglutinin (not shown). (A) Material eluted fromthe WGASepharose column was concentrated and run on a Superdex 200 HR 10/30 column, which yielded multiplepeaks. (B) Material in the peak fractions (shown by the shaded bar) was concentrated. Re-chromatography of pooledfractions from this central region gave a symmetrical peak, indicating homogeneity. From reference 18. Used withpermission. Copyright 2005 Elsevier Scientific Publication.

    -0.02

    0.00

    0.02

    0.04

    0.06

    0.10

    0.08

    0.12

    0.14

    5 10 15 20 25

    0.000

    0.001

    0.002

    0.004

    0.003

    0.005

    0.006

    5 10 15 20 25

    Volume (ml)

    Volume (ml)

    A

    B

    A280 nm

    A280 nm

  • 7/31/2019 Ge Seminar

    40/107

    40 Handbook 28-9095-31 AA

    Purity and homogeneity checkPurity checkAs with water-soluble proteins, SDS-PAGE is the most widespread method for assessing

    the purity of membrane proteins. Coomassie Blue, silver staining, or Deep PurpleTM (for

    fluorescence) can be used for detection. The Laemmli system (19) is commonly used. Some

    modifications may be necessary for membrane proteins, as outlined below. Suitable products

    are listed in the ordering information section.

    Boiling of the sample with SDS can cause aggregation of membrane proteins. As an

    alternative to boiling, incubation at 60C for 30 min or at 37C for 60 min are useful

    starting points for preparing the sample for SDS-PAGE. On the other hand, some

    membrane proteins are fully compatible with boiling and boiling may be required for

    complete solubilization with SDS.

    Membrane proteins frequently do not move according to the expected molecular

    weight in SDS-PAGE. They often move faster (i.e., appear smaller) possibly due to

    incomplete unfolding or due to binding more SDS per mass unit protein as compared

    with a water-soluble protein.

    SDS-PAGE Clean-Up kit has successfully been used to treat samples containing

    interfering detergent or that are too dilute for SDS-PAGE. With this kit, proteins are

    quantitatively precipitated while interfering substances remain in solution.

    Size homogeneity characterizationProtein aggregation is a common issue with membrane proteins. Aggregation often appears

    to be irreversible and it may occur slowly over time but also rapidly and unexpectedly

    with modest changes in ionic strength, pH, protein:detergent ratio and other factors. For

    membrane proteins, it is as important to keep track of aggregation as it is to monitor proteinactivity.

    Aggregation may not always be detected by SDS-PAGE since SDS solubilizes most

    aggregates. Gel filtration is the method of choice for rapid detection of aggregation and it can

    be applied under a wide variety of conditions. It is widely used as an efficient assay to assess

    the size homogeneity in purified membrane protein samples.

    Gel filtration allows detection of relatively small changes in the size of detergent-protein

    complexes when different detergents are compared. Gel filtration is often used to give an

    indication of the suitability of different detergents for a particular protein. Separation with

    gel filtration is thus an important tool for qualifying the membrane protein preparation forfurther analysis. As an example, gel filtration is very often used for assessing the suitability

    of different detergents for membrane protein crystallization. In some cases, however,


Recommended